IL-17E Human, HEK

Interleukin-17E Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Introduction to IL-17E Human, HEK

IL-17E (Interleukin-17E), also known as IL-25, is a glycosylated homodimeric cytokine belonging to the IL-17 family. It plays a critical role in Th2-mediated immune responses, eosinophil proliferation, and tissue inflammation. The "HEK" designation refers to its recombinant production in human embryonic kidney (HEK293) cells, which ensures proper post-translational modifications, such as glycosylation, essential for bioactivity . This recombinant protein is widely used in research for studying IL-17E signaling, receptor interactions, and therapeutic applications.

Signaling Pathway

IL-17E activates NF-κB and AP-1 pathways via:

  1. Receptor dimerization (IL-17RA/IL-17RB).

  2. Recruitment of ACT1 adaptor protein.

  3. TRAF6 ubiquitylation, triggering downstream kinase cascades .

Th2 Immune Response

IL-17E promotes eosinophilopoiesis and B-cell hyperplasia:

  • Eosinophilia: Transgenic mice overexpressing IL-17E exhibit 50-fold eosinophil increases, driven by IL-5 and eotaxin induction .

  • B-cell Proliferation: CD19+ B cells expand 2–10-fold in peripheral blood and lymph nodes, with elevated IgE, IgA, and IgG .

Inflammatory and Pathological Roles

  • Asthma and Allergy: Induces lung inflammation, mucus production, and airway hyperresponsiveness via Th2 cytokines (IL-4, IL-5, IL-13) .

  • Parasite Clearance: Essential for eradicating helminths (e.g., Trichuris muris) by enhancing Th2 responses .

HEK-Blue™ IL-17 Reporter Cells

These cells detect IL-17E bioactivity via SEAP (secreted embryonic alkaline phosphatase) production:

CytokineDetection RangeResponse
hIL-17E1–100 ng/mlNF-κB/AP-1 activation
mIL-17E1–100 ng/mlCross-reactivity
hIL-17A/F3–100 ng/mlLimited/no response

Adapted from InvivoGen HEK-Blue™ IL-17 specifications .

Transgenic Mouse Models

Mice overexpressing IL-17E exhibit:

  • Leukocytosis: 3-fold total leukocyte increase.

  • Splenomegaly and Lymphadenopathy: Due to eosinophil and lymphocyte infiltration .

  • Cytokine Dysregulation: Elevated IL-2, IL-4, IL-5, and G-CSF .

Therapeutic and Diagnostic Implications

  • Asthma and Autoimmunity: IL-17E antagonists may reduce eosinophil-driven inflammation .

  • Cancer and Infection: Dual roles in tumor promotion (via IL-17A/F) and pathogen clearance (IL-17E) .

Product Specs

Introduction

IL-17E, also known as IL-25, shares sequence similarity with IL-17. It activates NF-kappaB and stimulates the production of IL-8. Both IL-17E and IL-17B bind to the IL-17BR cytokine receptor. As a pro-inflammatory cytokine, IL-25 promotes Th2-type immune responses. Intracellular JNK, p38 MAPK, and NF-kappaB activity differentially regulate the upregulation of costimulation-induced IL-17E receptors and the release of cytokines and chemokines from IL-17E-treated, costimulated T helper cells. Blocking interleukin-25 prevents airway hyperresponsiveness, a key characteristic of clinical asthma. Innate effector eosinophils and basophils produce IL-25, which amplifies allergic inflammation by enhancing the survival and function of TSLP-DC-activated adaptive Th2 memory cells. In a transgenic mouse model, IL-25 overexpression leads to increased Th2 cytokine expression, growth retardation, jaundice, and multi-organ inflammation. IL-25 contributes to eosinophilic inflammation by acting on lung fibroblasts, highlighting its role in asthma pathophysiology. While IL-17E amplifies TH2 cell-mediated allergic airway inflammation, it does not directly induce it in vivo.

Description

Recombinant human IL-17E, produced in HEK293 cells, is a single, non-glycosylated polypeptide chain comprising 154 amino acids (33-177). It has a molecular weight of 17.8 kDa. The protein includes a C-terminal 6-histidine tag and is purified using proprietary chromatographic techniques.

Physical Appearance
Sterile, colorless solution.
Formulation

The IL-17E protein solution (0.5 mg/ml) is supplied in phosphate-buffered saline (pH 7.4) containing 20% glycerol.

Stability

For short-term storage (2-4 weeks), the product can be stored at 4°C. For long-term storage, freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for extended storage. Avoid repeated freeze-thaw cycles.

Purity

Purity is determined to be greater than 95% by SDS-PAGE analysis.

Biological Activity

Biological activity is assessed by the protein's ability to bind to human IL-17 RB in a functional ELISA assay.

Synonyms

interleukin-25 isoform 1, interleukin 25, IL17E, IL-17E, IL25, IL-25, interleukin-17E, interleukin-25.

Source

HEK293.

Amino Acid Sequence

DGSYSHWPSC CPSKGQDTSE ELLRWSTVPV PPLEPARPNR HPESCRASED GPLNSRAISP WRYELDRDLN RLPQDLYHAR CLCPHCVSLQ TGSHMDPRGN SELLYHNQTV FYRRPCHGEK GTHKGYCLER RLYRVSLACV CVRPRVMGHH HHHH

Q&A

What is the structural composition of recombinant human IL-17E?

Recombinant human IL-17E is a disulfide-linked homodimer. When produced in E. coli expression systems, it forms a non-glycosylated protein containing two 145-amino acid chains (with an N-terminal methionine for each chain, totaling 146 amino acids per chain) and has a predicted molecular mass of approximately 33.7 kDa . The human cell-expressed version encompasses amino acids Tyr33-Gly177 of the native protein . The disulfide linkages are critical for proper folding and biological activity.

How should researchers optimally reconstitute and store IL-17E for experimental use?

For optimal reconstitution of lyophilized IL-17E:

  • With carrier protein: Reconstitute at 100 μg/mL in 4 mM HCl containing at least 0.1% human or bovine serum albumin

  • Carrier-free versions: Reconstitute at 100 μg/mL in 4 mM HCl

Storage recommendations:

  • Store at -20°C upon receipt

  • Use a manual defrost freezer

  • Avoid repeated freeze-thaw cycles that can degrade the protein and reduce activity

  • Working aliquots should be prepared to minimize freeze-thaw cycles

What is the biological activity range of human IL-17E in functional assays?

Human IL-17E typically demonstrates biological activity in the range of 0.25-1.5 ng/mL in standard functional assays . In HEK-Blue IL-17 reporter systems, the detection range for human IL-17E is 1-100 ng/ml . The dose-response curve tends to plateau above 100 ng/ml, indicating receptor saturation. Researchers should construct full dose-response curves when first establishing assays with a new batch of IL-17E to determine optimal working concentrations.

What are the key signaling pathways activated by IL-17E?

IL-17E activates multiple signaling pathways:

  • NF-κB and AP-1 pathways: Primary pathways activated upon IL-17E binding to its receptor

  • JNK and p38 MAPK: These intracellular pathways differentially regulate the upregulation of IL-17E receptors and release of cytokines/chemokines

  • Promotes Th2 cytokine production: IL-4, IL-5, IL-13

  • Suppresses Th1/Th17 cytokines: IFN-gamma, IL-12, IL-23, IL-17A, IL-17F

How does IL-17E differ functionally from other IL-17 family members in immunological contexts?

While the IL-17 family generally promotes inflammatory responses, IL-17E stands apart with distinct immunoregulatory functions:

FeatureIL-17E/IL-25Other IL-17 Family Members
T-cell responsePromotes Th2-biased immunityPromote Th1/Th17-biased inflammation
Cytokine inductionInduces IL-4, IL-5, IL-13Induce IL-6, TNF-α, IL-1β
Role in diseaseMediates allergic reactions, protection against intestinal parasitesPromote autoimmunity, defense against extracellular pathogens
Impact on inflammationAllergic/eosinophilic inflammationNeutrophilic inflammation
Effect on autoimmunitySuppresses Th1/Th17-mediated autoimmunityExacerbate autoimmune pathology

This functional divergence makes IL-17E a therapeutic target in allergic disorders, while other family members are targeted in autoimmune conditions .

What protocol modifications are needed when studying IL-17E proteolytic activation by MMP-7?

When investigating MMP-7-mediated activation of IL-17E, researchers should consider:

  • Pre-incubation conditions: Recombinant IL-17E should be pre-incubated with purified MMP-7 at a 1:10 enzyme-to-substrate ratio for 1-2 hours at 37°C

  • Buffer composition: Use a buffer containing 10 mM CaCl₂ to ensure optimal MMP-7 activity

  • Verification of cleavage: Confirm processing by SDS-PAGE or Western blotting to detect the cleaved form

  • Functional comparison: Compare biological activity between cleaved and intact IL-17E by measuring:

    • Receptor binding affinity to IL-17RB

    • Induction of Th2 cytokines in target cells

This approach will help determine how proteolytic processing enhances IL-17E binding to IL-17RB and strengthens Th2 cytokine induction, as observed in airway allergic responses .

How can researchers address conflicting IL-17E expression data in tissue-specific studies?

Discrepancies in IL-17E expression data can arise from several methodological factors:

  • Detection technique sensitivity:

    • qPCR typically offers greater sensitivity than protein-based methods

    • Use digital droplet PCR for low-abundance transcripts

    • Validate with multiple antibody clones for protein detection

  • Cell isolation procedures:

    • Enzymatic digestion can alter surface receptor expression

    • Compare mechanical versus enzymatic isolation methods

    • Use gentle isolation protocols to preserve cytokine production capacity

  • Timing considerations:

    • IL-17E expression can be transient during immune responses

    • Perform time-course experiments (6h, 12h, 24h, 48h, 72h)

    • Consider diurnal variations in expression

  • Cross-validation approaches:

    • Triangulate findings using orthogonal methods (e.g., RNA-seq, proteomics, and functional assays)

    • Single-cell approaches can resolve heterogeneous expression patterns

    • In situ techniques preserve spatial context

When conflicting data emerge, systematic comparison of methodologies and detailed reporting of experimental conditions can help resolve apparent contradictions .

What is the optimal protocol for using HEK-Blue IL-17 cells to detect IL-17E bioactivity?

For optimal detection of IL-17E bioactivity using HEK-Blue IL-17 reporter cells:

Day 1:

  • Prepare HEK-Blue IL-17 cell suspension at ~280,000 cells/ml in test medium

  • Add 20 μl of IL-17E sample per well in a flat-bottom 96-well plate

  • Include positive control (recombinant human IL-17A at 10 ng/ml final concentration) and negative control (recombinant human TNF-α at 10 ng/ml)

  • Add 180 μl of cell suspension (~50,000 cells) per well

  • Incubate overnight at 37°C in 5% CO₂

Day 2:

  • Prepare QUANTI-Blue Solution according to manufacturer's instructions

  • Transfer 20 μl of induced cell supernatant to a new flat-bottom 96-well plate

  • Add 180 μl of resuspended QUANTI-Blue Solution

  • Incubate at 37°C for 30 min to 3 hours

  • Measure SEAP levels using a spectrophotometer at 620-655 nm

The detection range for human IL-17E is 1-100 ng/ml, with a typical dose-response curve showing sensitivity comparable to that of IL-17A .

How can researchers optimize HEK-Blue IL-17 cells for high-throughput screening of IL-17E inhibitors?

For adapting HEK-Blue IL-17 cells to high-throughput screening of IL-17E inhibitors:

  • Assay miniaturization:

    • Reduce volumes to 384-well format (10 μl cells + 5 μl compound + 5 μl IL-17E)

    • Maintain cell density at 50,000 cells per well

    • Use automated liquid handling for consistent results

  • Screening optimization:

    • Run Z'-factor determination using known inhibitors

    • Establish staggered addition protocol (pre-incubate compounds with IL-17E for 30 min)

    • Include concentration-response curves of reference inhibitors

  • Data normalization:

    • Include both positive (maximal stimulation) and negative (no stimulation) controls on each plate

    • Calculate percent inhibition relative to controls

    • Apply robust statistical methods (e.g., B-score) to account for plate position effects

  • Counter-screening:

    • Test hits against alternative activators of NF-κB/AP-1 pathways

    • Evaluate cytotoxicity using parallel viability assays

    • Confirm specificity by testing against other IL-17 family members

This approach enables efficient screening of large compound libraries while minimizing false positives and identifying selective IL-17E inhibitors .

What methodological approaches can address cross-reactivity when studying both human and mouse IL-17E in the same system?

When working with both human and mouse IL-17E in the same experimental system:

  • Receptor expression analysis:

    • HEK-Blue IL-17 cells respond to both human and mouse IL-17E

    • Characterize relative expression levels of IL-17RA and IL-17RB receptors by flow cytometry

    • Consider species-specific blocking antibodies to isolate responses

  • Species-specific dose-response assessment:

    • Determine EC₅₀ values for both human and mouse IL-17E

    • Human IL-17E typically shows higher potency in human cells

    • Quantify cross-reactivity ratios to calibrate mixed-species experiments

  • Selective inhibition strategies:

    • Use species-selective blocking antibodies

    • Design species-specific siRNA knockdown of receptor components

    • Consider CRISPR editing to create species-selective reporter cells

  • Data interpretation:

    • Always run parallel species-specific controls

    • Account for differences in receptor binding kinetics

    • Be aware that HEK-Blue IL-17 cells show stronger responses to human IL-17E than mouse IL-17F or IL-17A

This systematic approach allows researchers to dissect species-specific contributions in complex biological systems.

How should researchers design experiments to study IL-17E's differential effects on various cell types?

When investigating IL-17E's cell type-specific effects:

  • Target cell panel preparation:

    • Include epithelial cells (airway, intestinal, skin)

    • Test multiple immune cell types (T cells, eosinophils, basophils, dendritic cells)

    • Compare primary cells with relevant cell lines

    • Consider tissue-resident vs. circulating cell populations

  • Expression verification:

    • Confirm IL-17 receptor expression (IL-17RA/IL-17RB heterodimer) on each cell type

    • Quantify receptor levels by flow cytometry or qPCR

    • Assess expression of downstream signaling components (ACT1, TRAF6)

  • Multiparameter response measurement:

    • Cytokine/chemokine production (ELISA, multiplex assays)

    • Transcriptional changes (RNA-seq, targeted qPCR panels)

    • Signaling pathway activation (phospho-flow, Western blot for NF-κB, AP-1, MAPK)

    • Functional changes (migration, proliferation, survival)

  • Context-dependent modulation:

    • Test IL-17E effects in various inflammatory milieus

    • Examine synergy with allergic mediators (TSLP, IL-33)

    • Compare effects in homeostatic vs. inflammatory conditions

This comprehensive approach will reveal the tissue- and cell-specific impact of IL-17E across multiple biological contexts .

What are the methodological considerations for studying IL-17E in airway inflammation models?

For investigating IL-17E in airway inflammation models:

  • Sample processing protocol optimization:

    • Bronchoalveolar lavage (BAL): Process immediately and use protease inhibitors

    • Lung tissue: Standardize dissociation methods to preserve cell viability

    • Epithelial brushings: Optimize RNA/protein extraction from limited material

  • Timing considerations:

    • Early phase (0-6h): Focus on epithelial activation and initial cytokine release

    • Intermediate phase (24-48h): Measure immune cell recruitment and activation

    • Late phase (72h+): Assess tissue remodeling and chronic inflammation markers

  • Cell-specific analysis:

    • Flow cytometric identification of IL-17E-producing cells

    • Single-cell RNA-seq to capture heterogeneous responses

    • In situ hybridization to maintain spatial context of expression

    • Cell-specific knockout models to determine cellular sources

  • Functional readouts:

    • Airway hyperresponsiveness measurements (methacholine challenge)

    • Mucus production (PAS staining, MUC5AC quantification)

    • Eosinophil and Th2 cell infiltration

    • Local cytokine profiles (IL-4, IL-5, IL-13)

These methodological considerations will help researchers accurately characterize IL-17E's role in allergic airway inflammation and asthma pathophysiology .

How can researchers effectively study the therapeutic potential of IL-17E antagonism?

To investigate IL-17E antagonism as a therapeutic strategy:

  • Antagonist selection and characterization:

    • Neutralizing antibodies against IL-17E

    • Soluble receptor constructs (IL-17RB-Fc)

    • Small molecule inhibitors of IL-17E/receptor interaction

    • Verify specific binding and neutralizing capacity in vitro before in vivo studies

  • Preclinical model selection:

    • Acute vs. chronic allergen models

    • House dust mite vs. OVA-induced inflammation

    • Models with established epithelial barrier dysfunction

    • Humanized mouse models for increased translational relevance

  • Intervention timing optimization:

    • Prophylactic (before allergen challenge)

    • Early intervention (at symptom onset)

    • Therapeutic (during established disease)

    • Comparative efficacy at different disease stages

  • Comprehensive outcome assessment:

    • Primary outcomes: AHR, inflammation scores, symptom indices

    • Mechanistic outcomes: Th2 cytokine levels, ILC2 activation

    • Safety parameters: Impact on anti-helminth immunity, barrier integrity

    • Comparison with standard of care (corticosteroids, anti-IL-4/13)

This systematic approach will provide critical insights into the therapeutic window, efficacy parameters, and potential limitations of IL-17E antagonism as a treatment strategy for allergic diseases .

What are common technical challenges when working with recombinant IL-17E and their solutions?

When working with recombinant IL-17E, researchers may encounter several challenges:

ChallengePotential CausesSolutions
Loss of activityProtein aggregation, Improper storage, Freeze-thaw cyclesReconstitute with carrier protein (0.1% BSA/HSA), Prepare single-use aliquots, Use freshly thawed protein
Batch-to-batch variabilityExpression system differences, Purification methods, Protein modificationsCompare activity with reference standard, Use consistent supplier, Validate each batch in functional assays
Non-specific bindingProtein stickiness, Plastic tube adsorptionUse low-binding tubes, Add carrier protein, Handle at appropriate concentration
Interfering contaminantsEndotoxin contamination, Residual host proteinsUse endotoxin-tested preparations, Consider source (E. coli vs. human cell-expressed)
Precipitation upon reconstitutionImproper buffer, Excessive concentrationFollow manufacturer recommendations (4 mM HCl), Reconstitute at suggested concentration (100 μg/mL) , Allow complete dissolution at room temperature

Careful handling and quality control testing can mitigate most of these technical challenges.

How can researchers troubleshoot inconsistent responses in HEK-Blue IL-17 reporter assays?

For troubleshooting inconsistent HEK-Blue IL-17 reporter responses:

  • Cell health and culture conditions:

    • Check cell viability (>90% viable cells required)

    • Maintain proper cell density (avoid overconfluence)

    • Ensure appropriate passage number (confirmed stable for 20 passages)

    • Verify selection antibiotic maintenance (Blasticidin, Hygromycin B, Zeocin)

  • Assay execution variables:

    • Standardize incubation times (overnight stimulation)

    • Maintain consistent temperature (37°C) and CO₂ levels (5%)

    • Use freshly prepared QUANTI-Blue Solution

    • Control plate edge effects (use buffer in outer wells)

  • Signal detection optimization:

    • Verify spectrophotometer calibration

    • Optimize reading wavelength (620-655 nm)

    • Ensure appropriate integration time

    • Consider kinetic reading to determine optimal timepoint

  • Experimental controls:

    • Include positive control (hIL-17A at 10 ng/ml)

    • Include negative control (TNF-α at 10 ng/ml)

    • Run full dose-response curves

    • Test for receptor expression levels periodically

These systematic troubleshooting approaches can identify and address sources of variability in HEK-Blue IL-17 reporter assays .

What technical considerations should be addressed when analyzing IL-17E expression in complex tissue samples?

Analyzing IL-17E expression in complex tissues presents several technical challenges:

  • Sample preparation optimization:

    • Fresh samples provide most reliable results

    • Standardize time from collection to processing

    • Use RNase inhibitors for RNA analysis

    • Consider mild fixation methods that preserve epitopes

  • Detection method selection:

    • For mRNA: RNAscope offers cellular resolution with high sensitivity

    • For protein: Multiplex IHC with tyramide signal amplification

    • Single-cell approaches for heterogeneous tissues

    • Consider laser capture microdissection for region-specific analysis

  • Specificity controls:

    • Include isotype controls for antibody-based methods

    • Use IL-17E knockout tissue as negative control when available

    • Perform blocking experiments with recombinant protein

    • Validate findings with orthogonal methods

  • Quantification approaches:

    • Digital image analysis with machine learning algorithms

    • Colocalization with cell type-specific markers

    • Normalization to appropriate housekeeping genes/proteins

    • Relative quantification against standard curves

These considerations help ensure accurate detection and quantification of IL-17E in complex tissue environments where expression may be heterogeneous and often at low levels .

How can researchers effectively study IL-17E in the context of the gut-lung axis?

To investigate IL-17E's role in gut-lung immunological cross-talk:

  • Dual-site sampling strategy:

    • Coordinate intestinal and lung tissue collection

    • Analyze matched BAL and intestinal lavage fluids

    • Include draining lymph nodes from both compartments

    • Consider time-course sampling for migration studies

  • Microbiome influence assessment:

    • Compare germ-free, specific pathogen-free, and conventionally housed animals

    • Perform selective microbiota depletion using targeted antibiotics

    • Analyze metabolite profiles alongside IL-17E levels

    • Consider fecal microbiota transfer experiments

  • Cell trafficking studies:

    • Use photoconvertible protein systems (Kaede mice) to track cell migration

    • Employ adoptive transfer of labeled IL-17E-responsive cells

    • Analyze shared lymphoid populations between compartments

    • Monitor inflammatory cell recruitment patterns

  • Experimental perturbation approaches:

    • Compare airway vs. intestinal helminth infections

    • Study dual-site allergen challenges

    • Investigate how intestinal barrier disruption affects pulmonary IL-17E responses

    • Test compartment-specific IL-17E neutralization

These approaches help dissect the bidirectional communication between gut and lung immune systems, where IL-17E serves as a critical mediator in type 2 inflammation across mucosal barriers .

What are the methodological considerations for studying IL-17E in the context of epithelial barrier function?

For investigating IL-17E's impact on epithelial barrier integrity:

  • Barrier function measurement techniques:

    • Transepithelial electrical resistance (TEER) for tight junction integrity

    • FITC-dextran permeability assays with multiple molecular weights

    • Immunofluorescence analysis of junction proteins (claudins, occludin, ZO-1)

    • In vivo barrier assessment using serum biomarkers of permeability

  • Epithelial model selection:

    • Primary vs. immortalized epithelial cells

    • Air-liquid interface cultures for respiratory epithelium

    • Organoid models for three-dimensional architecture

    • Co-culture systems with immune cells

  • Molecular mechanisms investigation:

    • Junction protein expression analysis (qPCR, Western blot)

    • Cytoskeletal organization assessment (F-actin staining)

    • Mucin production quantification (MUC5AC, MUC2)

    • Antimicrobial peptide expression

  • Context-dependent modulation:

    • Examine IL-17E effects under basal vs. inflammatory conditions

    • Test barrier recovery after mechanical or chemical disruption

    • Compare acute vs. chronic IL-17E exposure

    • Investigate synergy with other epithelial-active cytokines (IL-13, TSLP)

These methodological approaches allow comprehensive evaluation of how IL-17E influences epithelial barrier function, which may contribute to its role in allergic inflammation and anti-helminth immunity .

How can transcriptomic approaches be optimized to study IL-17E-specific gene signatures?

For optimizing transcriptomic analysis of IL-17E-specific signatures:

  • Experimental design considerations:

    • Include multiple timepoints (early: 2-6h, intermediate: 12-24h, late: 48-72h)

    • Dose-response series to identify threshold-dependent genes

    • Compare with other IL-17 family members to identify unique signatures

    • Include IL-17E antagonism conditions for validation

  • Cell type optimization:

    • Use FACS-sorted primary cells when possible

    • Compare responses across different IL-17 receptor-expressing cells

    • Consider single-cell RNA-seq for heterogeneous populations

    • Include reference cell types for comparative analysis

  • Bioinformatic analysis pipeline:

    • Pathway enrichment focusing on Th2 inflammation and tissue remodeling

    • Transcription factor binding site analysis for key regulators

    • Integration with epigenomic data (ATAC-seq, ChIP-seq)

    • Network analysis to identify gene regulatory hubs

    • Cross-reference with asthma and allergy GWAS datasets

  • Validation approaches:

    • qPCR confirmation of key signature genes

    • Protein-level validation by Western blot or flow cytometry

    • Functional studies of identified targets

    • In vivo confirmation in relevant disease models

This comprehensive approach enables identification of robust IL-17E-specific transcriptional signatures that distinguish its function from other IL-17 family members and provide insights into its unique role in type 2 immune responses .

Product Science Overview

Introduction

Interleukin-17E (IL-17E), also known as Interleukin-25 (IL-25), is a member of the IL-17 cytokine family. This family comprises six members, all sharing a conserved cysteine-knot structure but differing at the N-terminus . IL-17E is a proinflammatory cytokine that plays a significant role in immune responses and inflammation .

Structure and Expression

IL-17E is secreted by type 2 helper T cells (Th2) and mast cells . It binds to the IL-17RB receptor, stimulating the secretion of proinflammatory cytokines such as interleukin-8 (IL-8) and activating nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) . The recombinant form of IL-17E, expressed in Human Embryonic Kidney (HEK) 293 cells, is used for various research and therapeutic purposes .

Biological Functions

IL-17E is involved in promoting inflammatory responses. It acts by recruiting immune cells like monocytes and neutrophils to the site of inflammation . This cytokine is known to work in concert with other proinflammatory cytokines such as tumor necrosis factor (TNF) and interleukin-1 (IL-1) . The activation of IL-17 signaling pathways is often observed in the pathogenesis of various autoimmune disorders, including psoriasis .

Therapeutic Potential

Research has shown that IL-17E has potential antitumor efficacy . Its ability to induce strong inflammatory responses makes it a candidate for cancer immunotherapy. Additionally, IL-17E’s role in autoimmune diseases suggests that targeting this cytokine could be beneficial in treating conditions like rheumatoid arthritis and multiple sclerosis .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.